Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
bioRxiv ; 2023 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-37781595

RESUMO

In cancer associated cachexia (CAC), white adipose tissue undergoes morphofunctional and inflammatory changes that lead to tissue dysfunction and remodeling. In addition to metabolic changes in white adipose tissues (WAT), adipose tissue atrophy has been implicated in several clinical complications and poor prognoses associated with cachexia. Adipocyte atrophy may be associated with increased beige remodeling in human CAC as evidenced by the "beige remodeling" observed in preclinical models of CAC. Even though beige remodeling is associated with CAC-induced WAT dysfunction, there are still some open questions regarding their cellular origins. In this study, we investigated the development of beige remodeling in CAC from a broader perspective. In addition, we used a grading system to identify the scAT as being affected by mice weight loss early and intensely. Using different in vitro and ex-vivo techniques, we demonstrated that Lewis LLC1 cells can induce a switch from white to beige adipocytes, which is specific to this type of tumor cell. During the more advanced stages of CAC, beige adipocytes are mainly formed from the transdifferentiation of cells. According to our results, humanizing the CAC classification system is an efficient approach to defining the onset of the syndrome in a more homogeneous manner. Pathological beige remodeling occurred early in the disease course and exhibited phenotypic characteristics specific to LLC cells' secretomes. Developing therapeutic strategies that recruit beige adipocytes in vivo may be better guided by an understanding of the cellular origins of beige adipocytes emitted by CAC.

2.
Cell Mol Bioeng ; 16(1): 23-39, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36660589

RESUMO

Introduction: Obesity is associated with increased breast cancer incidence, recurrence, and mortality. Adipocytes and adipose-derived stem cells (ASCs), two resident cell types in adipose tissue, accelerate the early stages of breast cancer progression. It remains unclear whether obesity plays a role in the subsequent escape of malignant breast cancer cells into the local circulation. Methods: We engineered models of human breast tumors with adipose stroma that exhibited different obesity-specific alterations. We used these models to assess the invasion and escape of breast cancer cells into an empty, blind-ended cavity (as a mimic of a lymphatic vessel) for up to sixteen days. Results: Lean and obese donor-derived adipose stroma hastened escape to similar extents. Moreover, a hypertrophic adipose stroma did not affect the rate of adipose-induced escape. When admixed directly into the model tumors, lean and obese donor-derived ASCs hastened escape similarly. Conclusions: This study demonstrates that the presence of adipose cells, independently of the obesity status of the adipose tissue donor, hastens the escape of human breast cancer cells in multiple models of obesity-associated breast cancer. Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-022-00750-y.

3.
Life Sci Alliance ; 5(5)2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35181634

RESUMO

Adipose tissue fibrosis is regulated by the chronic and progressive metabolic imbalance caused by differences in caloric intake and energy expenditure. By exploring the cellular heterogeneity within fibrotic adipose tissue, we demonstrate that early adipocyte progenitor cells expressing both platelet-derived growth factor receptor (PDGFR) α and ß are the major contributors to extracellular matrix deposition. We show that the fibrotic program is promoted by senescent macrophages. These macrophages were enriched in the fibrotic stroma and exhibit a distinct expression profile. Furthermore, we demonstrate that these cells display a blunted phagocytotic capacity and acquire a senescence-associated secretory phenotype. Finally, we determined that osteopontin, which was expressed by senescent macrophages in the fibrotic environment promoted progenitor cell proliferation, fibrotic gene expression, and inhibited adipogenesis. Our work reveals that obesity promotes macrophage senescence and provides a conceptual framework for the discovery of rational therapeutic targets for metabolic and inflammatory disease associated with obesity.


Assuntos
Adipócitos , Tecido Adiposo , Adipócitos/metabolismo , Tecido Adiposo/patologia , Fibrose , Humanos , Macrófagos/metabolismo , Obesidade/metabolismo
4.
Sci Signal ; 14(710): eabj2807, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34813359

RESUMO

Obesity and metabolic diseases, such as insulin resistance and type 2 diabetes (T2D), are associated with metastatic breast cancer in postmenopausal women. Here, we investigated the critical cellular and molecular factors behind this link. We found that primary human adipocytes shed extracellular vesicles, specifically exosomes, that induced the expression of genes associated with epithelial-to-mesenchymal transition (EMT) and cancer stem­like cell (CSC) traits in cocultured breast cancer cell lines. Transcription of these genes was further increased in cells exposed to exosomes shed from T2D patient­derived adipocytes or insulin-resistant adipocytes and required the epigenetic reader proteins BRD2 and BRD4 in recipient cells. The thrombospondin family protein TSP5, which is associated with cancer, was more abundant in exosomes from T2D or insulin-resistant adipocytes and partially contributed to EMT in recipient cells. Bioinformatic analysis of breast cancer patient tissue showed that greater coexpression of COMP (which encodes TSP5) and BRD2 or BRD3 correlated with poorer prognosis, specifically decreased distant metastasis­free survival. Our findings reveal a mechanism of exosome-mediated cross-talk between metabolically abnormal adipocytes and breast cancer cells that may promote tumor aggressiveness in patients with T2D.


Assuntos
Neoplasias da Mama , Diabetes Mellitus Tipo 2 , Exossomos , Adipócitos , Mama , Feminino , Humanos
5.
J Vis Exp ; (167)2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33522508

RESUMO

Cancer cachexia (CC) presents itself as a syndrome with multiple manifestations, causing a marked multi-organ metabolic imbalance. Recently, cachectic wasting has been proposed to be stimulated by several inflammatory mediators, which may disrupt the integrative physiology of adipose tissues and other tissues such as the brain and muscle. In this scenario, the tumor can survive at the host's expense. In recent clinical research, the intensity of depletion of the different fat deposits has been negatively correlated with the patient's survival outcome. Studies have also shown that various metabolic disorders can alter white adipose tissue (WAT) remodeling, especially in the early stages of cachexia development. WAT dysfunction resulting from tissue remodeling is a contributor to overall cachexia, with the main modifications in WAT consisting of morpho-functional changes, increased adipocyte lipolysis, accumulation of immune cells, reduction of adipogenesis, changes in progenitor cell population, and the increase of "niches" containing beige/brite cells. To study the various facets of cachexia-induced WAT remodeling, particularly the changes progenitor cells and beige remodeling, two-dimensional (2D) culture has been the first option for in vitro studies. However, this approach does not adequately summarize WAT complexity. Improved assays for the reconstruction of functional AT ex vivo help the comprehension of physiological interactions between the distinct cell populations. This protocol describes an efficient three-dimensional (3D) printing tissue culture system based on magnetic nanoparticles. The protocol is optimized for investigating WAT remodeling induced by cachexia induced factors (CIFs). The results show that a 3D culture is an appropriate tool for studying WAT modeling ex vivo and may be useful for functional screens to identify bioactive molecules for individual adipose cell populations applications and aid the discovery of WAT-based cell anticachectic therapy.


Assuntos
Adipócitos/patologia , Tecido Adiposo Branco/patologia , Caquexia/patologia , Técnicas de Cultura de Células/métodos , Modelos Biológicos , Adipócitos/metabolismo , Animais , Carcinoma Pulmonar de Lewis/patologia , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Humanos , Camundongos Endogâmicos C57BL , Nanopartículas/química , Perilipina-1/metabolismo , Esferoides Celulares/patologia , Células Estromais/patologia , Proteína Desacopladora 1/metabolismo
6.
Sci Rep ; 8(1): 18024, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30575787

RESUMO

Cancer-induced cachexia, characterized by systemic inflammation, body weight loss, adipose tissue (AT) remodeling and muscle wasting, is a malignant metabolic syndrome with undefined etiology. Here, we show that both genetic ablation and pharmacological inhibition of TLR4 were able to attenuate the main clinical markers of cachexia in mice bearing Lewis lung carcinoma (LLC). AT remodelling was not found in LLC tumor-bearing (TB) TLR4-/- mice due to reduced macrophage infiltration and adipocyte atrophy. TLR4-/- mice were also resistant to cold-induced browning of subcutaneous AT (scAT). Importantly, pharmacological inhibition of TLR4 (Atorvastatin) reproduced the main protective effect against AT remodeling found in TLR4-/- TB mice. Moreover, the treatment was effective in prolonging survival and attenuating tumor mass growth when compared to non-treated-TB animals. Furthermore, tumor-induced elevation of circulating pro-inflammatory cytokines was similarly abolished in both genetic ablation and pharmacological inhibition of TLR4. These data suggest that TLR4 is a critical mediator and a promising target for novel anti-cachexia therapies.


Assuntos
Tecido Adiposo/metabolismo , Caquexia/genética , Caquexia/mortalidade , Neoplasias/genética , Neoplasias/mortalidade , Receptor 4 Toll-Like/genética , Células 3T3-L1 , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/patologia , Adiposidade/efeitos dos fármacos , Adiposidade/genética , Animais , Atorvastatina/farmacologia , Caquexia/etiologia , Caquexia/metabolismo , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/mortalidade , Carcinoma Pulmonar de Lewis/patologia , Modelos Animais de Doenças , Deleção de Genes , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/complicações , Neoplasias/metabolismo , Análise de Sobrevida , Síndrome , Receptor 4 Toll-Like/antagonistas & inibidores , Células Tumorais Cultivadas
7.
Nat Commun ; 9(1): 1023, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29523786

RESUMO

Whereas white adipose tissue depots contribute to the development of metabolic diseases, brown and beige adipose tissue has beneficial metabolic effects. Here we show that CDK6 regulates beige adipocyte formation. We demonstrate that mice lacking the CDK6 protein or its kinase domain (K43M) exhibit significant increases beige cell formation, enhanced energy expenditure, better glucose tolerance, and improved insulin sensitivity, and are more resistant to high-fat diet-induced obesity. Re-expression of CDK6 in Cdk6 -/- mature or precursor cells, or ablation of RUNX1 in K43M mature or precursor cells, reverses these phenotypes. Furthermore, RUNX1 positively regulates the expression of Ucp-1 and Pgc1α by binding to proximal promoter regions. Our findings indicate that CDK6 kinase activity negatively regulates the conversion of fat-storing cells into fat-burning cells by suppressing RUNX1, and suggest that CDK6 may be a therapeutic target for the treatment of obesity and related metabolic diseases.


Assuntos
Tecido Adiposo Marrom/fisiologia , Tecido Adiposo Branco/fisiologia , Subunidade alfa 2 de Fator de Ligação ao Core/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/metabolismo , Regulação da Expressão Gênica , Adipócitos/citologia , Animais , Composição Corporal , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Cruzamentos Genéticos , Quinase 6 Dependente de Ciclina/genética , Dieta Hiperlipídica , Feminino , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Masculino , Doenças Metabólicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Consumo de Oxigênio , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fenótipo , Proteína Desacopladora 1/metabolismo
8.
Mol Metab ; 5(10): 970-979, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27689009

RESUMO

OBJECTIVE: Arising from common progenitors in the bone marrow, adipogenesis and osteogenesis are closely associated yet mutually exclusive during bone marrow mesenchymal stem cell (BMSC) development. Previous studies have shown that morphological changes can affect the early commitment of pluripotent BMSCs to the adipose versus osteoblastic lineage via modulation of RhoA activity. The RhoA pathway regulates actin polymerization to promote the incorporation of globular actin (G-actin) into filamentous actin (F-actin). In doing so, myocardin-related transcription factors (MRTFs) dissociate from bound G-actin and enter the nucleus to co-activate serum response factor (SRF) target gene expression. In this study, we investigated whether MRTFA/SRF is acting downstream of the RhoA pathway to regulate BMSC commitment in mice. METHODS: The effects of knocking out MRTFA on skeletal homeostasis was studied in MRTFA KO mice using micro-CT, QPCR and western blot assays. To determine how MRTFA affects the mechanisms regulating BMSC fate decisions, primary bone marrow stromal cells from WT and MRTFA KO mice as well as C3H10T1/2 cell lines were analyzed in vitro. RESULTS: Global MRTFA KO mice have lower whole body weight, shorter femoral and tibial lengths as well as significantly decreased bone mass in their femurs. BMSCs isolated from the KO mice show increased adipogenesis and reduced osteogenesis when compared to WT littermates. KO mice, particularly females, develop osteopenia with age, and this was enhanced by a high fat diet. Over-expression of MRTFA or SRF enhances osteogenesis in CH310T1/2 cell lines. Sca1(+), CD45(-) cells from KO marrow express lower amounts of smooth muscle actin (SMA) and TAZ/YAP target genes compared to WT counterparts. CONCLUSION: This study identified MRTFA as a novel regulator of skeletal homeostasis by regulating the balance between adipogenic and osteogenic differentiation of BMSCs. We propose that MRTFA promotes the osteogenic activity of TAZ/YAP by maintaining SMA production in BMSCs.

9.
PLoS One ; 10(3): e0122660, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25807446

RESUMO

Cachexia is a multifactorial syndrome characterized by profound involuntary weight loss, fat depletion, skeletal muscle wasting, and asthenia; all symptoms are not entirely attributable to inadequate nutritional intake. Adipose tissue and skeletal muscle loss during cancer cachexia development has been described systematically. The former was proposed to precede and be more rapid than the latter, which presents a means for the early detection of cachexia in cancer patients. Recently, pioglitazone (PGZ) was proposed to exhibit anti-cancer properties, including a reduction in insulin resistance and adipose tissue loss; nevertheless, few studies have evaluated its effect on survival. For greater insight into a potential anti-cachectic effect due to PGZ, 8-week-old male Wistar rats were subcutaneously inoculated with 1 mL (2×107) of Walker 256 tumor cells. The animals were randomly assigned to two experimental groups: TC (tumor + saline-control) and TP5 (tumor + PGZ/5 mg). Body weight, food ingestion and tumor growth were measured at baseline and after removal of tumor on days 7, 14 and 26. Samples from different visceral adipose tissue (AT) depots were collected on days 7 and 14 and stored at -80o C (5 to 7 animals per day/group). The PGZ treatment showed an increase in the survival average of 27.3% (P< 0.01) when compared to TC. It was also associated with enhanced body mass preservation (40.7 and 56.3%, p< 0.01) on day 14 and 26 compared with the TC group. The treatment also reduced the final tumor mass (53.4%, p<0.05) and anorexia compared with the TC group during late-stage cachexia. The retroperitoneal AT (RPAT) mass was preserved on day 7 compared with the TC group during the same experimental period. Such effect also demonstrates inverse relationship with tumor growth, on day 14. Gene expression of PPAR-γ, adiponectin, LPL and C/EBP-α from cachectic rats was upregulated after PGZ. Glucose uptake from adipocyte cells (RPAT) was entirely re-established due to PGZ treatment. Taken together, the results demonstrate beneficial effects of PGZ treatment at both the early and final stages of cachexia.


Assuntos
Carcinoma 256 de Walker/tratamento farmacológico , Tiazolidinedionas/uso terapêutico , Adiponectina/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Caquexia/patologia , Carcinoma 256 de Walker/mortalidade , Carcinoma 256 de Walker/patologia , Linhagem Celular Tumoral , Ingestão de Alimentos/efeitos dos fármacos , Masculino , PPAR gama/metabolismo , Pioglitazona , Ratos , Ratos Wistar , Taxa de Sobrevida , Tiazolidinedionas/farmacologia , Transplante Homólogo
10.
Cell ; 160(1-2): 105-18, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25579684

RESUMO

Adipose tissue is an essential regulator of metabolic homeostasis. In contrast with white adipose tissue, which stores excess energy in the form of triglycerides, brown adipose tissue is thermogenic, dissipating energy as heat via the unique expression of the mitochondrial uncoupling protein UCP1. A subset of UCP1+ adipocytes develops within white adipose tissue in response to physiological stimuli; however, the developmental origin of these "brite" or "beige" adipocytes is unclear. Here, we report the identification of a BMP7-ROCK signaling axis regulating beige adipocyte formation via control of the G-actin-regulated transcriptional coactivator myocardin-related transcription factor A, MRTFA. White adipose tissue from MRTFA(-/-) mice contains more multilocular adipocytes and expresses enhanced levels of brown-selective proteins, including UCP1. MRTFA(-/-) mice also show improved metabolic profiles and protection from diet-induced obesity and insulin resistance. Our study hence unravels a central pathway driving the development of physiologically functional beige adipocytes.


Assuntos
Transativadores/metabolismo , Adipogenia , Animais , Proteína Morfogenética Óssea 7/metabolismo , Dieta , Metabolismo Energético , Resistência à Insulina , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Transativadores/genética , Fator de Crescimento Transformador beta1/metabolismo
11.
Cell ; 150(3): 620-32, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863012

RESUMO

Brown adipose tissue (BAT) can disperse stored energy as heat. Promoting BAT-like features in white adipose (WAT) is an attractive, if elusive, therapeutic approach to staunch the current obesity epidemic. Here we report that gain of function of the NAD-dependent deacetylase SirT1 or loss of function of its endogenous inhibitor Deleted in breast cancer-1 (Dbc1) promote "browning" of WAT by deacetylating peroxisome proliferator-activated receptor (Ppar)-γ on Lys268 and Lys293. SirT1-dependent deacetylation of Lys268 and Lys293 is required to recruit the BAT program coactivator Prdm16 to Pparγ, leading to selective induction of BAT genes and repression of visceral WAT genes associated with insulin resistance. An acetylation-defective Pparγ mutant induces a brown phenotype in white adipocytes, whereas an acetylated mimetic fails to induce "brown" genes but retains the ability to activate "white" genes. We propose that SirT1-dependent Pparγ deacetylation is a form of selective Pparγ modulation of potential therapeutic import.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , PPAR gama/metabolismo , Sirtuína 1/metabolismo , Células 3T3 , Acetilação , Adulto , Sequência de Aminoácidos , Animais , Células Cultivadas , Metabolismo Energético , Feminino , Humanos , Resistência à Insulina , Ligantes , Lisina/análise , Lisina/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Obesidade/complicações , Obesidade/metabolismo , PPAR gama/química , Resveratrol , Alinhamento de Sequência , Sirtuína 1/química , Sirtuína 1/genética , Estilbenos/farmacologia , Termogênese , Tiazolidinedionas/farmacologia
12.
J Biol Chem ; 287(22): 18351-8, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22493496

RESUMO

Obese white adipose tissue is hypoxic but is incapable of inducing compensatory angiogenesis. Brown adipose tissue is highly vascularized, facilitating delivery of nutrients to brown adipocytes for heat production. In this study, we investigated the mechanisms by which white and brown adipocytes respond to hypoxia. Brown adipocytes produced lower amounts of hypoxia-inducible factor 1α (HIF-1α) than white adipocytes in response to low O(2) but induced higher levels of hypoxia-associated genes. The response of white adipocytes to hypoxia required HIF-1α, but its presence alone was incapable of inducing target gene expression under normoxic conditions. In addition to the HIF-1α targets, hypoxia also induced many inflammatory genes. Exposure of white adipocytes to a peroxisome proliferator-activated receptor γ (PPARγ) ligand (troglitazone) attenuated induction of these genes but enhanced expression of the HIF-1α targets. Knockdown of PPARγ in mature white adipocytes prevented the usual robust induction of HIF-1α targets in response to hypoxia. Similarly, knockdown of PPARγ coactivator (PGC) 1ß in PGC-1α-deficient brown adipocytes eliminated their response to hypoxia. These data demonstrate that the response of white adipocytes requires HIF-1α but also depends on PPARγ in white cells and the PPARγ cofactors PGC-1α and PGC-1ß in brown cells.


Assuntos
Adipócitos/citologia , Tecido Adiposo Marrom/citologia , Hipóxia Celular , PPAR gama/fisiologia , Transativadores/fisiologia , Células 3T3 , Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição
13.
J Lipid Res ; 52(9): 1693-701, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21743036

RESUMO

Recent studies have established SIRT1 as an important regulator of lipid metabolism, although the mechanism of its action at the molecular level has not been revealed. Here, we show that knockdown of SIRT1 with the help of small hairpin RNA decreases basal and isoproterenol-stimulated lipolysis in cultured adipocytes. This effect is attributed, at least in part, to the suppression of the rate-limiting lipolytic enzyme, adipose triglyceride lipase (ATGL), at the level of transcription. Mechanistically, SIRT1 controls acetylation status and functional activity of FoxO1 that directly binds to the ATGL promoter and regulates ATGL gene transcription. We have also found that depletion of SIRT1 decreases AMP-dependent protein kinase (AMPK) activity in adipocytes. To determine the input of AMPK in regulation of lipolysis, we have established a stable adipose cell line that expresses a dominant-negative α1 catalytic subunit of AMPK under the control of the inducible TET-OFF lentiviral expression vector. Reduction of AMPK activity does not have a significant effect on the rates of lipolysis in this cell model. We conclude, therefore, that SIRT1 controls ATGL transcription primarily by deacetylating FoxO1.


Assuntos
Adipócitos/enzimologia , Fatores de Transcrição Forkhead/metabolismo , Regulação Enzimológica da Expressão Gênica , Lipase/metabolismo , Metabolismo dos Lipídeos , Lipólise/fisiologia , Sirtuína 1/metabolismo , Células 3T3-L1 , Adenilato Quinase/metabolismo , Adipócitos/citologia , Adipócitos/fisiologia , Animais , Regulação para Baixo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Lipase/genética , Camundongos , PPAR gama/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Sirtuína 1/genética
14.
J Biol Chem ; 282(36): 26046-56, 2007 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-17611194

RESUMO

Recent reports demonstrate that peroxisome proliferator-activated receptor gamma (PPARgamma), a member of the nuclear receptor superfamily, acts as a repressor of type I collagen synthesis. Our data demonstrate that exogenously expressed PPARgamma down-regulates collagen expression in a dose-responsive manner in human lung fibroblast cells. Silencing PPARgamma using lentiviruses expressing short hairpin RNAs partially reverses interferon-gamma (IFN-gamma)-induced repression and activates collagen mRNA levels. Previous studies indicate that IFN-gamma represses collagen gene expression and induces major histocompatibility complex II (MHC II) expression by activating the formation of a regulatory factor for X-box 5 (RFX5) complex with class II transactivator (CIITA). This report demonstrates that PPARgamma is within the RFX5.CIITA complex as judged by co-immunoprecipitation and DNA affinity precipitation studies. Most importantly, occupancy of PPARgamma on the collagen transcription start site and MHC II promoter increases with IFN-gamma treatment. The PPARgamma agonist, troglitazone, sensitizes the cells to IFN-gamma treatment by increasing recruitment of PPARgamma to collagen gene while repressing collagen expression, and these effects are blocked by the PPARgamma antagonist T0070907. PPARgamma may mediate IFN-gamma-stimulated collagen transcription down-regulation and MHC II up-regulation by interacting with CIITA as well as regulating CIITA expression. Therefore, PPARgamma is a critical target for investigations into therapeutics of diseases involving extracellular matrix remodeling and the immune response.


Assuntos
Colágeno Tipo I/biossíntese , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/fisiologia , Fibroblastos/metabolismo , Pulmão/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , PPAR gama/metabolismo , Transativadores/metabolismo , Antineoplásicos/farmacologia , Antivirais/farmacologia , Benzamidas/farmacologia , Linhagem Celular , Cromanos/farmacologia , Colágeno Tipo I/genética , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Inativação Gênica , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Interferon gama/farmacologia , Lentivirus , Pulmão/citologia , Complexos Multiproteicos/genética , Proteínas Nucleares/genética , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , PPAR gama/genética , Piridinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Transcrição de Fator Regulador X , Tiazolidinedionas/farmacologia , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Troglitazona , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
15.
Mol Cell Biol ; 26(15): 5827-37, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847334

RESUMO

Studies have demonstrated cross talk between beta-catenin and peroxisome proliferator-activated receptor gamma (PPARgamma) signaling pathways. Specifically, activation of PPARgamma induces the proteasomal degradation of beta-catenin in cells that express an adenomatous polyposis coli-containing destruction complex. In contrast, oncogenic beta-catenin is resistant to such degradation and inhibits the expression of PPARgamma target genes. In the present studies, we demonstrate a functional interaction between beta-catenin and PPARgamma that involves the T-cell factor (TCF)/lymphocyte enhancer factor (LEF) binding domain of beta-catenin and a catenin binding domain (CBD) within PPARgamma. Mutation of K312 and K435 in the TCF/LEF binding domain of an oncogenic beta-catenin (S37A) significantly reduces its ability to interact with and inhibit the activity of PPARgamma. Furthermore, these mutations render S37A beta-catenin susceptible to proteasomal degradation in response to activation of PPARgamma. Mutation of F372 within the CBD (helices 7 and 8) of PPARgamma disrupts its binding to beta-catenin and significantly reduces the ability of PPARgamma to induce the proteasomal degradation of beta-catenin. We suggest that in normal cells, PPARgamma can function to suppress tumorigenesis and/or Wnt signaling by targeting phosphorylated beta-catenin to the proteasome through a process involving its CBD. In contrast, oncogenic beta-catenin resists proteasomal degradation by inhibiting PPARgamma activity, which requires its TCF/LEF binding domain.


Assuntos
PPAR gama/metabolismo , Transdução de Sinais/fisiologia , beta Catenina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , DNA/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Lisina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , PPAR gama/química , PPAR gama/genética , Fenilalanina/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Transcrição TCF/metabolismo , beta Catenina/química , beta Catenina/genética
16.
J Biol Chem ; 281(32): 23025-33, 2006 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16754681

RESUMO

Preadipocyte differentiation capacity declines between middle and old age. Expression of the adipogenic transcription factors, CCAAT/enhancer-binding protein (C/EBP) alpha and peroxisome proliferator-activated receptor gamma (PPARgamma), is lower in differentiating preadipocytes from old than young animals, although no age-related changes occur in C/EBPbeta mRNA, which is upstream of C/EBPalpha and PPARgamma. C/EBPbeta-liver-enriched inhibitory protein (C/EBPbeta-LIP), a truncated C/EBPbeta isoform that is a dominant inhibitor of differentiation, increases with aging in rat fat tissue and preadipocytes. CUG triplet repeat-binding protein-1 (CUGBP1) binds to C/EBPbeta mRNA, increasing C/EBPbeta-LIP translation. Abundance and nucleotide binding activity of CUGBP1 increased with aging in preadipocytes. CUGBP1 overexpression in preadipocytes from young animals increased C/EBPbeta-LIP and impaired adipogenesis. Decreasing CUGBP1 in preadipocytes from old rats by RNA interference reduced C/EBPbeta-LIP abundance and promoted adipogenesis. Tumor necrosis factor-alpha, levels of which are elevated in fat tissue with aging, increased CUGBP1 protein, CUGBP1 binding activity, and C/EBPbeta-LIP in preadipocytes from young rats. Thus, CUGBP1 contributes to regulation of adipogenesis in primary preadipocytes and is responsive to tumor necrosis factor-alpha. With aging, preadipocyte CUGBP1 abundance and activity increases, resulting in enhanced translation of the C/EBPbeta-LIP isoform, thereby blocking effects of adipogenic transcription factors, predisposing preadipocytes from old animals to resist adipogenesis. Altered translational processing, possibly related to changes in cytokine milieu and activation of stress responses, may contribute to changes in progenitor differentiation and tissue function with aging.


Assuntos
Adipócitos/metabolismo , Adipogenia , Envelhecimento , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação da Expressão Gênica , Nucleotídeos/química , PPAR gama/metabolismo , Proteínas de Ligação a RNA/fisiologia , Animais , Proteínas CELF1 , Diferenciação Celular , Ligação Proteica , Interferência de RNA , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo
17.
Am J Physiol Endocrinol Metab ; 291(1): E175-81, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16464908

RESUMO

Thiazolidinediones (TZDs) are insulin-sensitizing agents used in the treatment of type 2 diabetes. A widely held view is that their action is secondary to transcriptional events that occur when TZDs bind to the nuclear receptor PPARgamma in the adipocyte and stimulate adipogenesis. It has been proposed that this increases insulin sensitivity, at least in part, by increasing the expression and release of adiponectin, an adipokine that activates the fuel-sensing enzyme AMP-activated protein kinase (AMPK). In this study, we report that TZDs also acutely activate AMPK in skeletal muscle and other tissues by a mechanism that is likely independent of PPARgamma-regulated gene transcription. Thus incubation of isolated rat EDL muscles in medium containing 5 microM troglitazone for 15 min (too brief to be attributable to transcription) significantly increased pAMPK and pACC. At a concentration of 100 microM, troglitazone maximally increased these parameters and caused twofold increases in 2-deoxy-d-glucose uptake and the oxidation of exogenous [(14)C]palmitate. Time course studies revealed that troglitazone-induced increases in pAMPK and pACC abundance at 15 min were paralleled by an increase in the AMP-to-ATP ratio and that by 60 min all of these parameters had returned to baseline values. Increases in pAMPK and pACC were also observed in skeletal muscle, liver, and adipose tissue in intact rats 15 min after the administration of a single dose of troglitazone (10 mg/kg, ip). Likewise, troglitazone and another TZD, pioglitazone, caused rapid increases in pAMPK and pACC of equal magnitude in Swiss 3T3 fibroblasts with and without sufficient PPARgamma to mediate the expression of target genes. The results indicate that TZDs can act within minutes to activate AMPK in mammalian tissues. They suggest that this effect is associated with a change in cellular energy state and that it is not dependent on PPARgamma-mediated gene transcription.


Assuntos
Cromanos/farmacologia , Hipoglicemiantes/farmacologia , Complexos Multienzimáticos/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Tiazolidinedionas/farmacologia , Proteínas Quinases Ativadas por AMP , Acetil-CoA Carboxilase/metabolismo , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Ativação Enzimática , Ácidos Graxos/metabolismo , Glucose/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Músculo Esquelético/metabolismo , Oxirredução , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Células Swiss 3T3 , Troglitazona
18.
Biochem Biophys Res Commun ; 340(3): 845-51, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16376852

RESUMO

Western blot analysis of 3T3-L1 adipocyte proteins using an anti-C/EBPalpha antibody detected a 24kD polypeptide in addition to the expected 42 and 30kD isoforms of C/EBPalpha. Mass spectrometric sequencing of the protein following its purification by HPLC and preparative 2D gel electrophoresis identified it as glutathione S-transferase zeta/maleylacetoacetate isomerase (GSTzeta/MAAI). Expression of GSTzeta/MAAI mRNA and protein was induced during the terminal phase of adipogenesis in 3T3-L1 preadipocytes. Ectopic expression of PPARgamma2 in NIH-3T3 fibroblasts exposed to insulin and troglitazone-induced perilipin production, but was incapable of activating GSTzeta/MAAI unless C/EBPalpha was also expressed. Similarly, ectopic expression of C/EBPalpha in PPARgamma +/- or PPARgamma -/- MEFs demonstrated that the C/EBPalpha-dependent induction of GSTzeta/MAAI production was dependent on expression of endogenous PPARgamma. These data suggest a role for GSTzeta/MAAI in mature adipocytes that may be responsive to the thiazolidinedione class of insulin sensitizing PPARgamma ligands.


Assuntos
Adipócitos/enzimologia , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Fibroblastos/enzimologia , Regulação Enzimológica da Expressão Gênica , Glutationa Transferase/biossíntese , cis-trans-Isomerases/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Antioxidantes/metabolismo , Western Blotting , Diferenciação Celular , Células Cultivadas , Cromatografia Líquida de Alta Pressão , DNA Complementar/metabolismo , Eletroforese em Gel Bidimensional , Fibroblastos/citologia , Vetores Genéticos , Glutationa Transferase/metabolismo , Ligantes , Metabolismo dos Lipídeos , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , PPAR gama/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Plasmídeos/metabolismo , Isoformas de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
19.
Am J Physiol Endocrinol Metab ; 288(6): E1195-205, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15644454

RESUMO

Peroxisome proliferator-activated receptor-gamma (PPARgamma) is considered to be one of the master regulators of adipocyte differentiation. PPARgamma2 is abundantly expressed in mature adipocytes and is elevated in the livers of animals that develop fatty livers. The aim of this study was to determine the ability of PPARgamma2 to induce lipid accumulation in hepatocytes and to delineate molecular mechanisms driving this process. The hepatic cell line AML-12 was used to generate a cell line stably expressing PPARgamma2. Oil Red O staining revealed that PPARgamma2 induces lipid accumulation in hepatocytes. This phenotype is accompanied by a selective upregulation of several adipogenic and lipogenic genes including adipose differentiation-related protein (ADRP), adipocyte fatty acid-binding protein 4, sterol regulatory element-binding protein-1 (SREBP-1), fatty acid synthase (FAS), and acetyl-CoA carboxylase, genes whose expression levels are known to increase in steatotic livers of ob/ob mice. Furthermore, the PPARgamma2-regulated induction of both SREBP-1 and FAS parallels an increase in de novo triacylglycerol synthesis in hepatocytes. Triacylglycerol synthesis and lipid accumulation are further enhanced by culturing hepatocytes with troglitazone in the absence of exogenous lipids. These results correspond with an increase in the lipid droplet protein, ADRP, and the data demonstrate that ADRP functions to coat lipid droplets in hepatocytes as observed by confocal microscopy. Taken together, these observations propose a role for PPARgamma2 as an inducer of steatosis in hepatocytes and suggest that this phenomenon occurs through an induction of pathways regulating de novo lipid synthesis.


Assuntos
Fígado Gorduroso/metabolismo , Metabolismo dos Lipídeos , PPAR gama/metabolismo , Acetil-CoA Carboxilase/biossíntese , Acetil-CoA Carboxilase/genética , Animais , Compostos Azo/química , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas de Transporte/biossíntese , Proteínas de Transporte/genética , Cromanos/farmacologia , Proteínas de Ligação a DNA/genética , Ácido Graxo Sintases/biossíntese , Ácido Graxo Sintases/genética , Proteínas de Ligação a Ácido Graxo , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/fisiologia , Hipoglicemiantes/farmacologia , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Microscopia Confocal , PPAR gama/antagonistas & inibidores , PPAR gama/biossíntese , Perilipina-2 , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína de Ligação a Elemento Regulador de Esterol 1 , Tiazolidinedionas/farmacologia , Fatores de Transcrição/genética , Triglicerídeos/biossíntese , Troglitazona
20.
Mol Cell Biol ; 24(16): 7151-62, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15282314

RESUMO

Two families of transcription factors that play a major role in the development of adipocytes are the CCAAT/enhancer-binding proteins (C/EBPs) and the peroxisome proliferator-activated receptors (PPARs), in particular PPAR gamma. Ectopic expression of either C/EBP alpha or PPAR gamma in NIH 3T3 fibroblasts results in the conversion of these cells to adipocyte-like cells replete with fat droplets. NIH 3T3 cells ectopically expressing C/EBP alpha (NIH-C/EBP alpha) differentiate into adipocytes and exhibit insulin-stimulated glucose uptake, whereas NIH 3T3 cells ectopically expressing PPAR gamma (NIH-PPAR gamma) differentiate but do not exhibit any insulin-stimulated glucose uptake, nor do they express any C/EBP alpha. The reason for the lack of insulin-responsive glucose uptake in the NIH-PPAR gamma cells is their virtual lack of the insulin-responsive glucose transporter, Glut4. The NIH-PPAR gamma cells express functionally active components of the insulin receptor-signaling pathway (the insulin receptor, IRS-1, phosphatidylinositol 3-kinase, and Akt2) at levels comparable to those in responsive cell lines. They also express components of the insulin-sensitive vesicular transport machinery, namely, VAMP2, syntaxin-4, and IRAP, the last of these being the other marker of insulin-regulated vesicular traffic along with Glut4. Interestingly, the NIH-PPAR gamma cells show normal insulin-dependent translocation of IRAP and form an insulin-responsive vesicular compartment as assessed by cell surface biotinylation and sucrose velocity gradient analysis, respectively. Moreover, expression of a Glut4-myc construct in the NIH-PPAR gamma cells results in its insulin-dependent translocation to the plasma membrane as assessed by immunofluorescence and Western blot analysis. Based on these data, we conclude that major role of C/EBP alpha in the context of the NIH-PPAR gamma cells is to regulate Glut4 expression. The differentiated cells possess a large insulin-sensitive vesicular compartment with negligible Glut4, and Glut4 translocation can be reconstituted on expression of this transporter.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Vesículas Citoplasmáticas/metabolismo , Insulina/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Adipócitos/citologia , Adipócitos/fisiologia , Aminopeptidases/metabolismo , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Diferenciação Celular/fisiologia , Membrana Celular/metabolismo , Cistinil Aminopeptidase , Desoxiglucose/metabolismo , Fibroblastos/citologia , Fibroblastos/fisiologia , Transportador de Glucose Tipo 4 , Camundongos , Proteínas de Transporte de Monossacarídeos/genética , Células NIH 3T3 , Transporte Proteico/fisiologia , Receptor de Insulina/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA